MECHANISMS - PowerPoint PPT Presentation

1 / 69
About This Presentation
Title:

MECHANISMS

Description:

MECHANISMS – PowerPoint PPT presentation

Number of Views:213
Avg rating:3.0/5.0
Slides: 70
Provided by: Michae809
Category:
Tags: mechanisms | guus

less

Transcript and Presenter's Notes

Title: MECHANISMS


1
MECHANISMS
2
(No Transcript)
3
(No Transcript)
4
MechanismsStep 1 - Toxicant Delivery
  • Absorption
  • Presystemic Elimination
  • Traverse epithelial barriers and reach blood
    capillaries by diffusing through cells.
  • Rate prop to C prop to exposure and dissolution
    and surface area of absorbing surface.
  • Lipid-soluble chemicals absorbed more readily
    than H2O-soluble chemicals.
  • GIT and lungs are major players (e.g., ethanol,
    morphine), leads to first-pass elimination
    decreases toxicity of chemical before they can
    ever harm the rest of the body.

5
  • Distribution Towards Target
  • Enhanced by
  • Distribution Away from Target
  • Enhanced by
  • Porosity of capillary endothelium due to large
    fenestrae that permit passage of even
    protein-bound xenobiotics.
  • Specialized membrane transport (e.g.,
    ATPase-hydrolysis, MPTP.
  • Accumulation in cell organelles (e.g., MPTP
    accumulates in the mitochondria of dopamine
    neurons).
  • Binding to plasma proteins (e.g., DDT, TCDD bind
    high-MW proteins in plasma, preventing their
    escape from capillaries by diffusion).
  • Specialized barriers (e.g., BBB endothelial cells
    lack fenestrae, have tight junctions, oocyte
    surrounded by granulosa cells, spermatogenic
    cells surrounded by Sertoli cells all prevent
    hydrophillic substances from gaining entry).
    However, lipophillic substances can gain entry.
  • Distribution to storage sites (e.g., bone,
    kidneys, liver, fat) protect target tissues from
    toxicants

6
  • Reversible intracellular binding (e.g., by
    binding to the pigment melanin, PAHs accumulate
    in melanin-containing cells also, release of
    melanin-bound toxicants contributes to the
    retinal toxicity assoc. with chlorpromazine and
    chloroquine injury to SN neurons by MPTP, and
    induction of melanoma by polycyclic aromatics).
  • Association with intracellular binding proteins
    (e.g., metallothionen binds Cd in the event of
    acute Cd intoxication).
  • Export from cells. Intracellular toxicants may
    be transported back into the extracellular space
    (e.g., brain capillary endothelial cells, which
    contain in their luminal membrane an
    ATP-dependent membrane transporter
    (P-glycoprotein) - contributes to the BBB.

7
  • Reabsorption
  • Excretion
  • High lipid solubility.
  • Many glucuronide conjugates.
  • Lipid-soluble compounds are reabsorbed by
    transcellular diffusion.
  • Removal of xenobiotics from the blood and their
    return to the external environment
  • Renal glomeruli hydrostatically filter small
    molecules (lt 60 kDal) through their pores
    (HOH-soluble and ionized).
  • Transporters in hepatocytes and proximal tubular
    cells are specialized for the secretion of highly
    hydrophillic organic acids and bases eliminated
    in bile and urine high pH-dependency

8
  • Toxification
  • Detoxification
  • Sometimes, it is the chemical itself is directly
    toxic (e.g., HCN, CO, strong acids and bases,
    nicotine).
  • Most often, however, toxification renders
    xenobiotics and sometimes, autocoids (O2 and NO)
    indiscriminantly reactive towards endogenous
    molecules
  • Electophiles
  • (e.g., chloroform overhead)
  • Free Radicals (next 2 slides)
  • Nucleophiles - Relatively rare (e.g.,
    dihalomethandes undergo oxidative dehalogenation
    to yield CO).
  • Redox-active reactiants
  •  
  • Eliminates the ultimate toxicant or prevents its
    formation.
  • Detoxification of toxicants with no functional
    groups (e.g., benzene or toluene) undergoes Phase
    I and Phase II reactions - readily excreted as
    hydrophillic organic acids.
  • Detoxification of Nucleophiles - Phase II
    reactions prevent peroxidase-catalyzed conversion
    of the nucleophiles to FRs and biotransformation
    of phenols, catechols and hydroquinones to
    electrophiles.
  • Detoxification of Electrophiles - Commonly occurs
    with the thiol nucleophile GSH, facilitated by
    GSH-S-transferase.
  • Detoxification of FRs. Generation of FRs must be
    undone by various enzymes - See handouts for
    generation of superoxide anion and for its
    elimination by potentially 3 different enzymes.
  • Detoxification of protein toxins - many venoms
    contain intramolecular disulfid bonds, which can
    be reduced by an endogenous dithiol protein
    thioredoxin.

9
(No Transcript)
10
(No Transcript)
11
(No Transcript)
12
(No Transcript)
13
  • In summary, the most reactive metabolites are
    electron-deficient molecules and molecular
    fragments such as neutral or cationic free
    radicals.
  • Other FRs with an extra electron cause damage by
    yielding the neutral OH after the formation and
    subsequent homolytic cleavage of HOOH.
  • When Detoxification Fails
  • Toxicants may overwhelm detox processes, leading
    to exhaustion of the detox enzymes, consumption
    of substrates, or depletion of antioxidants such
    as GSH, Asc. acid, and alpha-tocopherol.
  • Occasionally, a reactive toxicant inactivates a
    detoxicating enzyme (e.g., ONOO- incapacitates
    Mn-SOD, which would normally counteract ONOO-
    formation.
  • Some conjugation reactions can be reversed (e.g.,
    bladder carcinogen 2-naphthylamine is
    hydroxylated and glucuronidated in liver and
    excreted into urine while in bladder, the
    glucuronide is hydrolyzed and the released
    arylhydroxylamine is protonated and dehydrated to
    the reactive electrophile arylnitrenium ion).
  • Detoxification can sometimes generate potentially
    harmful by-products, such as GSH thyl radical and
    GSH disulfide, which are produced during the
    detoxification of FRs.

14
(No Transcript)
15
(No Transcript)
16
(No Transcript)
17
Step 2 Reaction of the Ultimate Toxicant with
the Target Molecule
  • I. Attributes of the target molecules
  • Reactivity
  • Accessibility
  • Critical function
  • Reactive metabolites that cannot find appropriate
    endogenous molecules in close proximity to their
    site of formation may diffuse until they
    encounter such reactants.
  • Not all chemical targets contribute to harmful
    effects (e.g., CO has deleterious consequences
    when it binds the heme in Hb, but not when it
    binds the heme in cytochrome P-450.
  • II. Reaction Types
  • 1. Noncovalent binding ionic, H, van der
    Waals interaction of toxicants with receptors,
    ion channels, some enzymes, intracellular
    receptors (e.g., strychnine binds using these
    interactions when binding the GABA receptor in
    the spinal cord TCDD to the Ah receptor
    saxitoxin to Na channels)
  • Such forces are also responsible for
    intercalation of chemicals such as acridine
    orange into the DNA double helix

18
2. Covelent binding Irreversible. Thus greatly
and permanently alters endogenous molecules.
E.g., elecrophillic reactions with nucleophillic
atoms neutral FRs to DNA. 3. Hydrogen
Abstraction Fenton Reaction (Fig. 3-4) and lipid
peroxidation (Fig. 3-9). 4. Electron
Transfer Oxidation of the Fe in Hb from II to
III produces methemoglobinemia. 5. Enzymatic
Reactions a few toxins act like enzymes in
specific target proteins (e.g., ricin inhibits
protein synthesis by hydrolyzing ribosomes).
19
(No Transcript)
20
(No Transcript)
21
(No Transcript)
22
  • III. Outcomes Effects of Toxicants on Target
    Molecules
  • Dysfunction of Target Molecules some toxicants
    activate endogenous molecules, such as mimicking
    endogenous ligands (e.g., morphine to opiate
    receptors phorbol esters to protein kinase C).
  • More commonly, many toxins inhibit function of
    endogenous molecules e.g., atropine, curare, or
    strychnine inhibit Ach receptor-medieated nerve
    transmission.
  • Also, interference with template function in DNA
    e.g., covalent binding of aflatoxin 8,9-oxide to
    N7 of G results in base-pairing of the G with A,
    rather, than with C, leading to incorrect codon
    and a point mutation.
  • Destruction of Target Molecules Cross-linking
    of endogenous molecules e.g., OH radicals can
    cross-link macromolecules into reactive
    electrophiles, such as protein carbonyls.
  • Also, lipid peroxidation (See Fig. 3-9).
  • Neoantigen Formation Occasionally, covalent
    binding of xenobiotics or their metabolites to
    macromolecules results in Ab production in some
    individuals e.g., CYP-450 biotransforms
    halothane to an electrophile (trifluoroacetyl
    Cl), which binds as a hapten to various
    microsomal and cell surface proteins in the
    liver, thereby inducing an immune response (Ab
    production).

23
(No Transcript)
24
Step 3 Cellular Dysfunction and Resultant
Toxicities
  • I. Toxicant-Induced Cellular Dysregulation
  • A. Dysregulation of Gene Expression
  • Occurs at elements that are directly responsible
    for transcription, signal transduction pathways,
    synthesis, storage, or release of extracellular
    signaling molecules.
  • B. Dysregulation of Transcription
  •  
  • Xenobiotics may interact/promote/interfere with
    promotor regions, interfering with transcription
    factors (TFs).
  • e.g., many hormones, vitamins influence gene
    expression by binding to and activating TFs.
  • Xenobiotics may mimic the natural compounds
    (e.g., Cd2 may substitute for Zn2 at the
    metal-responsive element-binding TF.
  • e.g., estrogen acts as a mitogen normally in
    female reproductive organs but prolonged
    exposure induces tumor formation in these organs.
  • e.g, TCDD, Phenobarbital, and pregnenolone
    activate the AhR, thereby inducing CYP1A1

25
  • C. Dysregulation of Signal Transduction
  • Fig. 3-11 in (painstaking) detail NEXT SLIDE.
  • Xenobiotics may increase phosphorylation too much
    (e.g., TCDD-liganded AhR binds MAPK may
    contribute to TCDD-induced overexpression of
    kinase activity in liver.
  • Abberant phosphorylation or proteins may result
    not only from increased phosphorylation, but also
    from decreased phosphatase activity, such as is
    seen with various chemicals, oxidative stress,
    and UV radiation, which inhibit phosphatase
    activity
  • e.g., arsenite, tributyltin, and oxidants, such
    as HOOH, cause phosphorylation of the EGF
    receptor by interfering with the tyr phosphatase
    that would dephosphorylate this receptor.
  • e.g., PP2A dephosphorylates MAPK, keeping MAPK
    under control.
  • However, some naturally occurring toxins are
    extremely potent inhibitors of PP2A (e.g.,
    blue-green algae poison microsystin-LG and the
    dinoflagellate-derived okadeic acid), which are
    tumor-promotors in experimental animals exposed
    to prolonged low doses.

26
(No Transcript)
27
  • Acute high dose of microsystin-LG ? severe liver
    injury.
  • Acute high dose of okadeic acid ? diarrhetic
    shellfish poisoning.
  • But this may be due to hyperphosphorylation of
    proteins other than those involved in
    intracellular signaling (e.g., microfillaments)
    e.g., phosphorylation of tau in Alzheimers may
    be caused by too much FRs, oxidant production,
    tyr phosphatase inhibited?

28
  • D. Dysregulation of Extracellular Signal
    Production
  • Hormonal intereference (e.g., herbicide amitrole
    inhibit thyroid hormone production Phenobarbital
    enhance thyroid hormone elimination)
  •  
  • Both decrease thyroid hormone levels and increase
    the secretion of TSH because of the reduced
    feedback inhibition.
  • Another e.g., estrogens (e.g., xenoestrogen
    chlordecone) produce male testicular atrophy by
    means of feedback inhibition of gonadotropin
    secretion ? low sperm count.

29
  • E. Dysregulation of Ongoing cellular activity
  • Note the myriad agents that act on signaling
    systems for neurotransmitters and causing
    dysregulation of momentary activity of
    electrically excitable cells such as neurons and
    myocytes.
  • F. Dysregulation of the activity of other Cells
  • Many exocrine secretory cells are governed by
    muscarinic Ach receptors, e.g., lacrimation and
    salivation are due to organophosphate insecticide
    poisoning due to stimulation of these receptors.

30
  • II. Toxic Alteration of Cellular Maintenance
  • A. Impairment of Internal Cellular Maintenance
    Mechanisms of Toxic Cell Death
  • ATP depletion
  • Sustained rise in intracellular Ca2
  • Overproduction of ROS and RNS
  • Depletion of ATP. Recall Oxidative
    Phosphorylation mitochondrial function of ATP
    production, delivery of H in the form of NADH,
    formation of the H gradient, O2 delivery to the
    terminal electron-transport complex and the
    reduction of O2 to H2O.
  • Sustained rise in intracellular Ca2. Fig. 3-14.
    Influenced by 3 Mechanisms
  • High cytoplasmic Ca2 levels cause increased
    mitochondrial Ca2 uptake by the Ca2 uniporter,
    which uses the mitochondrial internal negative
    membrane potential as the driving force. The
    membrane potential, therefore, dissipates and ATP
    production ceases.
  • ATP synthesis is impaired by Ca2-induced
    oxidative injury to the inner mitochondrial
    membrane
  • (3) ATP consumption is increased due to the
    Ca2-ATPase working overtime to eliminate the
    excess Ca2.
  • Overproduction of ROS and RNS (recall Fig. 3-3
    and Fig. 3-4).

31
(No Transcript)
32
(No Transcript)
33
(No Transcript)
34
(No Transcript)
35
Interplay Among Primary Metabolic Cell Disorders
Spells Disaster For The Cell
  • Mito. Permeability Transfer (MPT) and the worst
    possible outcome Necrosis.
  • MPT ? permeability of the mitochondrial inner
    membrane
  • ? Mitochondrial Ca2 uptake
  • ? ??m
  • ? ROS, RNS
  • ? ATP
  • ? Pi, FFAs

36
  • Proteinaceous pore opens up and spans both
    mitochondrial membranes (pore size lt 1.5 kDal).
  • Causes free influx of Ca2, H2O, dissipation of
    the ??m, osmotic swelling, ? ATP synthesis,
    lysis.
  • Also, depolarization of inner mito. membranes
    ?ATP synthesis to operate in reverse, as in
    ATPase, hydrolyzing ATP.
  • Also, glycolysis may become compromised by
    insufficient ATP to supply ATP-requiring enzymes
    (e.g., PFK, hexokinase).
  • Then, oxidative and hydrolytic degredation of
    macromolecules and membranes and degredation of
    intracellular milieu ? cell lysis (necrosis).

37
Alternative Outcome of MPT Apoptosis
  • Necrosis Cell
  • Apoptotic Cell
  • Swells
  • Lysis
  • Chaotic, random sequence of events
  • Shrinks
  • Cyto and nuclear materials condense, break off
    into membrane-bound fragments (apoptotic bodies)
    that are phagocytosed.
  • Membranes bleb.
  • Ordered, cascade-like activation of catabolic
    processes that disassemble the cell.

38
(No Transcript)
39
(No Transcript)
40
ATP Availability Determine the Form of Cell Death
  • Common features of Necrosis and Apoptosis
  • Many xenobiotics (e.g., acetominophen, ocratoxin)
    cause both.
  • A. Apoptosis induced at low exposure levels and
    early after high exposure levels.
  • B. Necrosis later at higher exposure levels.
  • Similar metabolic disturbances, e.g., MPT.
  • Blockers of necrosis (e.g., Bcl2 over-expression
    will block apoptosis.

41
ATP Availability is Critical in Determining the
form of Cell Death
MPT
In few mito and cytochrome c are removed
by lysosomal autophagy
In all mito. ATP Severely depleted. Cannot
execute apoptotic program (requires
ATP). Cytolysis occurs before caspases
are activated.
In a few more mito autophagic mech are
overwelmed released cyt c initiates caspase and
activation and apoptosis.
42
(No Transcript)
43
Step 4 Repair or Disrepair
  • MOLECULAR REPAIR
  • Repair of proteins
  • Some Methods
  • Repair of Oxidized Hb (Met Hb) e- transfer from
    cyt. b5 from cyt b5 reductase (-NAHPH-dependent).
  • Chaperones e.g., heat-shock proteins refold
    altered proteins.
  • Proteosomes proteolytically degrade mutated
    protein.

44
  • Repair of Lipids.
  • NAPDH-dependent repair of peroxidized lipids
    using GSH peroxidase and reductase.
  • Repair of DNA.
  • Despite high reactivity with electrophiles and
    FRs, nuclear DNA is quite stable, partly because
    its packaged into chromatin, highly folded, and
    has several repair mechanisms. Mitochondrial
    DNA, however, is more prone to damage because it
    lacks histones and efficient repair mechanisms.

45
Some DNA-Damaging AgentsFour Major Categories
  • Direct-acting carcinogens intrinsically
    reactive and dont require metabolic activation
    by cellular enzymes to covalently interact with
    DNA e.g., N-methyl-N-nitrosourea,
    alkylsulfonates (methyl methanesulfonate),
    lactones (beta-propriolactone), and N and S
    mustards.
  •  Indirect-acting carcinogens require metabolic
    activation by cellular enzymes to form the
    ultimate carcinogenic species that covalently
    binds to DNA e.g., dimethylnitrosamine,
    benzoapyrene, 7,12-dimethylbenzaanthracene,
    aflatoxin B1, and 2-acetylaminofluorene.

46
Some DNA-Damaging AgentsFour Major Categories
(contd)
  • Radiation and oxidative damage can occur either
    indirectly or directly ionizing radiation to
    produce ds breaks or ionization of water to
    produce ROS/RNS that damage DNA bases. UV
    radiation also.
  • 4. Inorganic agents (e.g., As, Cr, Ni), but many
    mechanisms are unknown but 3 general types of
    genetic alterations can result

47
Some DNA-Damaging AgentsFour Major Categories
(contd)
  1. Gene mutations point- (bp-substitutions) and
    frame-shift mutations.
  2. Chromosome aberrations (gross rearrangements,
    deletions, duplications, inversions, and
    translocations).
  3. Aneuploidy, polyploidy.

48
(No Transcript)
49
(No Transcript)
50
  • Direct Repair DNA photolyase
  • Excision Repair Base/nucleotide excision,
    ligase, polymerase, PARP.
  • Recombinatorial Repair
  • --sister chromatid exchange.
  • --Occurs when the excision of a bulky adduct or
    an intrastrand pyrimidine dimer fails to occur
    before DNA replication begins.
  • --Cross-over and recombination.

51
  • CELLULAR REPAIR
  • Nonneural tissue divide to replace lost cells.
  • Neurons dont multiply.
  • PNS Macrophages and Schwann Cells
  • Phagocytosis
  • and
  • Growth factors and cytokines
  • facilitate transdifferentiation from
  • myelination mode to growth-
  • supporting mode.
  • CNS Lost neurons are compensated by
  • neighboring neurons.

52
  • TISSUE REPAIR
  • Tissue is composed of t components
  • cells and ECM. 2 Processes
  • Apoptosis Deleting damaged cells.
  • Proliferation Regeneration of (damaged)
    tissue.
  • Replacement of lost cells by
    mitosis.

53
  • Replacement of the ECM.
  • In liver stellate cells and adipocytes
  • (spaces of Disse)
  • ECM proteins
  • glycosaminoglycans
  • glycoprotein
  • proteoglycan glucoconjugates

54
  • When Repair Fails
  • Repair mechanisms are not absolute can overlook
    various lesions.
  • Repair can contribute to toxicity.
  • e.g., after chronic tissue injury when the
    repair process goes astray and leads to
    uncontrolled proliferation, rather than tissue
    remodeling. Such proliferation of cells may
    yield neoplasms, whereas overproduction of ECM
    results in fibrosis.

55
  • Toxicity Resulting from Disrepair
  • Tissue Necrosis.
  • e.g, lipid peroxidation can be repaired unless
    a-tocopherol is depleted ? cell injury progresses
    toward cell necrosis if molecular repair
    mechanisms are insufficient or the molecular
    damage is not readily reversible.
  • -- Fibrosis.
  • Pathological condition characterized by
    excessive deposition of abnormally composed ECM
    or too much ECM. E.g., chronic alcohol leading
    to hepatic fibrosis and cirrhosis.

56
  • --- Carcinogenesis
  • Chemical carcinogenesis involves insufficient
    function of various repair mechanisms including
  • (1) Failure of DNA repair.
  • (2) Failure of Apoptosis.
  • (3) Failure to terminate cell proliferation.

57
  • Failure of DNA repair Mutation is the
    initiating event in carcinogenesis.
  • DNA damage adduct formation
  • oxidative changes
  • strand breakage
  • Failure to repair lesion ? heritable alteration
    (mutation) in the daughter strands during
    replication. Mutations can reprogram cells for
    multiplication. Enhanced cell divisions ?
  • likelihood of mutations.

58
(No Transcript)
59
  • Proto-oncogenes.
  • Highly conserved genes encoding proteins that
    stimulate the progression of cells through the
    cell cycle.
  • Products are growth factors, GF receptors,
    intracellular signaling transducers, such as G
    proteins, protein kinases, and nuclear
    transcription factors.
  • Genetic carcinogens mutate proto-oncogenes ?
    Oncogene.
  • Constitutive sustained activation of the
    promotor region of the proto-oncogenem e.g.,
    TCDD or benzo(a)pyrene can activate the Ah
    receptor ? binds the promotor on Ras ?
    transactivation in perpetuity.

60
(No Transcript)
61
(No Transcript)
62
  • Tumour-Suppressor Genes
  • p53 Guardian of the genome because it
    eliminates cancer-prone cells from the
    replicative pool, counteracting neoplasmic
    transformation.
  • Mutations in p53 gene found in 50 of human
    tumors and in a variety of induced cancers.
  • Cells with no p53 are 106 times more likely to
    permit DNA amplification then are cells with a
    normal level of p53 suppressor protein.

63
  • Cooperation of Proto-oncogenes and Tumor
    Suppressor Genes in Carcinogenesis
  • The accumulation of genetic damage is due to
    mutant proto-oncogenes and mutant tumor
    suppressor genes.
  • ? This is the main driving force in the
    transformation of normal cells with controlled
    proliferative activity (normal mitosis) to
    malignant cells with uncontrolled proliferative
    activity (cancer cells, metastatic).
  • Because the normal number of cells in a tissue is
    regulated by a balance between mitosis and
    apoptosis, uncontrolled proliferation results
    from a disturbance of this balance.

64
(No Transcript)
65
(No Transcript)
66
(No Transcript)
67
  • (2) Failure of Apoptosis Promotion of mutation
    and clonal growth.
  • Apoptosis eliminates cells with damaged DNA,
    preventing mutation, which initiates
    carcinogenesis.
  • Inhibiting apoptosis facilitates mutation and
    clonal expansion of pre-neoplastic cells
  • e.g., phenobarbital.

68
  • (3) Failure to Terminate Replication.
  • A. Enhanced mitotic activity ? P(mutations).
  • G1 phase of cell cycle is shortened ? ? time
    used for DNA repair ?? P(mutations).
  • B. During increased proliferation,
    proto-oncogenes are over-expressed, which may
    cooperate with oncogene proteins to facilitate
    neoplastic transformation of cells. ? ? time for
    DNA methylation at C5 of C residues ? enhances
    gene expression and may result in over-expression
    of proto-oncogenes and oncogenes.

69
  • C. Cell-cell communication through gap
    junctions and intercellular adhesion through
    cadherins are temporarily disrupted during
    proliferation.
  • Lack of these junctions contributes to the
    invasiveness of tumor cells.
  • Several tumor promotors (phenobarbitol, phorbol
    esters) ? gap junctions and intercellular
    communication.
Write a Comment
User Comments (0)
About PowerShow.com